WO2022225602A1 - Plates-formes fluidiques pour tissus vascularisés perfusables avec infiltrations - Google Patents

Plates-formes fluidiques pour tissus vascularisés perfusables avec infiltrations Download PDF

Info

Publication number
WO2022225602A1
WO2022225602A1 PCT/US2022/017505 US2022017505W WO2022225602A1 WO 2022225602 A1 WO2022225602 A1 WO 2022225602A1 US 2022017505 W US2022017505 W US 2022017505W WO 2022225602 A1 WO2022225602 A1 WO 2022225602A1
Authority
WO
WIPO (PCT)
Prior art keywords
gel
cells
channel
microfluidic
ports
Prior art date
Application number
PCT/US2022/017505
Other languages
English (en)
Inventor
Roger Kamm
Huu Tuan NGUYEN
Sharon Wei Ling LEE
Original Assignee
Massachusetts Institute Of Technology
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Massachusetts Institute Of Technology filed Critical Massachusetts Institute Of Technology
Publication of WO2022225602A1 publication Critical patent/WO2022225602A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M23/00Constructional details, e.g. recesses, hinges
    • C12M23/02Form or structure of the vessel
    • C12M23/16Microfluidic devices; Capillary tubes
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0278Physical preservation processes
    • A01N1/0284Temperature processes, i.e. using a designated change in temperature over time
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M21/00Bioreactors or fermenters specially adapted for specific uses
    • C12M21/08Bioreactors or fermenters specially adapted for specific uses for producing artificial tissue or for ex-vivo cultivation of tissue
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/02Membranes; Filters
    • C12M25/04Membranes; Filters in combination with well or multiwell plates, i.e. culture inserts
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12MAPPARATUS FOR ENZYMOLOGY OR MICROBIOLOGY; APPARATUS FOR CULTURING MICROORGANISMS FOR PRODUCING BIOMASS, FOR GROWING CELLS OR FOR OBTAINING FERMENTATION OR METABOLIC PRODUCTS, i.e. BIOREACTORS OR FERMENTERS
    • C12M25/00Means for supporting, enclosing or fixing the microorganisms, e.g. immunocoatings
    • C12M25/14Scaffolds; Matrices
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/069Vascular Endothelial cells
    • C12N5/0691Vascular smooth muscle cells; 3D culture thereof, e.g. models of blood vessels
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0697Artificial constructs associating cells of different lineages, e.g. tissue equivalents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2513/003D culture
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/56Fibrin; Thrombin

Definitions

  • This invention is generally directed to microfluidic devices supporting tissue perfusion and vascularization.
  • Microvasculature has unique biological functions and physical properties, such as maintaining solute and water balance between the blood and tissue compartments, and responding to different deformations and stress fluctuations (Jain et a., Nat. Med., 9:685-693 (2003)).
  • organ-on-a-chip has been proposed to establish in vitro models that can mimic the microphysiological function and three-dimensional (3D) microstructure of human organs more accurately and specifically compared to the traditional two-dimensional (2D) cultures and animal models (Bhatia et al.,
  • vascularization of an organ-on-a-chip can also contribute to the establishment of organ- specific microenvironments and microphysiological function by constructing the microvascular with selective barrier function similar to that in vivo.
  • organ-on-a-chip systems Wang et al., Micromachines, 9, 493:1-26 (2018).
  • Microfluidic technologies have emerged as useful tools for the development of organs-on-a-chip, which can help control various aspects of the cellular microenvironment such as a different profile of fluid flow, gradient of various growth factors, and mechanical properties of versatile biomaterials.
  • Organic-on-a-chip models enable the development of 3D cultures of different cell types and allow detailed characterization of critical biological interactions (Boussommier-Calleja et al., Trends in Cancer. ⁇ , 2(1):6-19 (2016), Sontheimer- Phelps, et al., Nat Rev Cancer: 19:65-81 (2019)). These microfluidic chips have the capability to recapitulate a perfusable vascular network and deliver signaling molecules and immune cells using microfluidic flow (Chen et al., Nat Protoc:, 12(5):865-80 (2017)).
  • Spheroids are created by culturing human cells in 3D on a low adhesion well plate or in a gel.
  • Organoids are cells in 3D culture that are derived from a tissue, embryonic stem cells or induced pluripotent stem cells.
  • Animal and patient-derived organoids are typically ex vivo tissues that are obtained by dissection and surgery, respectively.
  • spheroids, organoids, and ex vivo tissues are better representations of the structure of an organ where each cell is in contact with other cells and incorporated into the extracellular matrix.
  • Integrating 3D structures such as spheroids, organoids, or ex vivo tissues into microfluidic systems not only recapitulates the physiological state of the cells within a body but also allows interactions between cells in a tissue and peripheral cells or circulating immune cells (Shelton et al. iScience. 24 (1), 101985 (2021)).
  • Vasculature-on-a-chip models are used to study the mechanism and interactions between different cells and the vasculature (Boussommier-Calleja et al., Biomaterials, 198:180-93 (2019)), drug transport, disease modeling, or cancer metastasis (Chen et al., Nat Protoc. ⁇ , 12(5):865-80 (2017)).
  • Vasculatures have been created using a predefined hollow gel wall coated with endothelial cells (Ayuso et al., Oncoimmunology, 8(3) (2019)), angiogenesis of endothelial cells grown as a monolayer, or vasculogenesis of endothelial cells suspended in a gel (Chen et al., Integr Biol Quant Biosci from Nano to Macro ;5( 10): 1262-71 (2013), Campisi et al., Biomaterials, 180:117-29 (2018)).
  • endothelial cells angiogenesis of endothelial cells grown as a monolayer
  • vasculogenesis of endothelial cells suspended in a gel Choen et al., Integr Biol Quant Biosci from Nano to Macro ;5( 10): 1262-71 (2013), Campisi et al., Biomaterials, 180:117-29 (2018).
  • perfusable vascular networks In order to study cell-vasculature interactions
  • vasculature structures based on the angiogenesis of an endothelial monolayer, have been formed before grafting a tumor spheroid from the top (Nashimoto et al., Biomaterials., 229:119547 (2020)).
  • endothelial cells form self-assembled vascular networks with the presence of spheroids or organoids or ex vivo tissues, which affect the network’s morphology, density, and connectivity.
  • tissue isolated from a patient can compete for nutrients with endothelial cells that are forming vasculatures. Insertion of tissue samples into a fully-formed microvasculature bed ensures the viability of freshly-isolated tissues or time- sensitive spheroid/organoid samples.
  • nutrients can be delivered to the tissue samples through flows inside the vasculatures.
  • inserting samples is by adding spheroids/ organoids/ tissue samples onto the top of a vasculature bed within an open-top microfluidic device (Vulto et al, US 2020/0063081 Al (2016), Paek et al., ACS Nano, 13(7):7627- 43 (2019)).
  • any transport of cells or molecules from vasculatures to spheroids/organoids happens in the vertical direction and useful imaging area at the interface between the vasculature and spheroids/organoids situates at the top of the vascular bed that has a typical thickness between 100-1000 pm (Chen et al., Nat Protoc., 12(5): 865-80 (2017), Paek et ak, ACS Nano, 13(7):7627-43 (2019), Campisi et ak, Biomaterials, 180:117-29 (2016)).
  • Immunotherapies constitute an expanding therapeutic armamentarium against various diseases, including cancer and viral infection. Due to the complex mechanism of immunotherapy, and oftentimes, their lack of cross-reactivity to murine counterparts of their intended targets, drug testing using animal models or conventional 2D human cell cultures are not sufficient to predict patient drug efficacy. Many in vitro models do not model both immune cell extravasation and chemotaxis migration steps (Paek et ak, ACS Nano 13(7):7627-43 (2019), Ayuso et ak, Oncoimmunology, 8(3) (2019)).
  • microfluidic platforms for forming and culturing perfusable pre-vascularized tissues with infiltrates ⁇
  • Microfluidic devices for forming perfusable vascularized tissues with one or more empty wells and infiltrates typically include at least three adjacent and parallel microchannels, wherein a central microchannel is typically a gel channel having cells forming microvascular networks and a central port therein for the introduction of organoids/spheroids/tissues, and the two adjacent microchannels are media channels.
  • the gel channel includes a first end and a second end and is separated from the media channels by phase guides.
  • the devices also include one or more ports positioned on a top surface of the gel channel and at a distance away from the first end and the second end of the gel channel.
  • the one or more ports are positioned in the top surface of the gel channel and, in one embodiment, at about the center of the gel channel.
  • the one or more ports may be positioned in the top surface of the gel channel and at a distance between about 1 mm and about 20 mm away from the first end and the second end.
  • the gel channel includes one port. In some embodiments, the gel channel includes more than one port.
  • the microfluidic devices are filled with a gel solution containing cells and/or one or more extracellular matrix (ECM) components.
  • the cells may be endothelial cells, stromal cells, smooth muscle cells, pericytes, fibroblasts, progenitor cells, or combinations thereof.
  • Exemplary ECM components include fibrous proteins such as collagen, fibrin, fibronectin, elastins, and laminin; hyaluronic acid; and proteoglycans. These cells form a vascularized network within the gel. Oxygenated culture media is passed through one of the adjacent media channels, so that the fluid and oxygen pass through the gel to nourish and maintain the vascular network, and into the second media channel.
  • the method of filling the devices with the gel solution includes forming holes within the gel solution below one or more ports of the device.
  • the gel channel After filling the gel channel with the gel solution, allowing it to gel, forming holes in the gel, and culturing, the gel channel typically includes a perfusable vascular network with one or more holes, each hole positioned below one of the one or more ports.
  • the formation of the hole surrounded by fully-formed microvasculatures is an important difference compared to previous in vitro tissue models.
  • One or more microfluidic devices may form microfluidic platforms with fluidic connections to fluid supply reservoirs.
  • each device has a fluidic connection to the fluid supply reservoir via the gel channel port and via the media.
  • the method typically includes depositing one or more cells, spheroids, organoids, ex vivo tissues, or mixes of cells in the one or more holes of the perfusable vascular network positioned on the top of the gel channel of the microfluidic device and culture for a period of time.
  • the deposited cells are cultured in the devices with a perfusable vascular network for between about 2 days and 10 days.
  • cells can be supplied through the media channels, then typically infiltrate into the deposited tissue masses in the gel channel to form perfusable tissue masses with infiltrates.
  • These methods are also useful in forming vascularized human tumors for immunotherapy drug screening.
  • the devices with perfusable vascular networks and open-top ports typically generate fluid flow which passes through the implanted cells or tissue toward the vasculature, mimicking the interstitial flow of tissue microenvironments.
  • Interstitial flow integration is key for the recruitment of immune cells into the organoid or spheroid.
  • Perfusable vascularized tissue masses can include infiltrates of at least three categories: (1) infiltrated cells that extravasate and migrate toward the tissue masses and enter the tissue masses; (2) cells that extravasate but do not move toward the tissue masses; and (3) cells that stay luminal.
  • the devices are useful for studying molecular and cell transports from vasculatures to tissues and vice-versa, the effects and/or effective dosage of therapeutic, prophylactic, and/or diagnostic agents, for assessing an immunological role or effect on vascular structures, and for characterizing cellular interactions between the sample and the vascular network.
  • the agent(s)e can be applied directly to the sample or vascular network through the entry/sample port in the gel channel.
  • the transport of the agents is characterized by real-time imaging of the tissues and used to compute pharmacokinetic parameters.
  • the effects of the agents can be assessed visually through the entry/sample port, by detecting and/or measuring cell viability, phenotype, cell migration or cell composition, cellular function (such as contraction of heart cells or production of insulin by islet cells) or changes in gene expression or products produced by the cells in response to the agent, or by other means of quantitating cells or cellular products or changes therein over time as a function of the agent.
  • angiogenesis can be assessed by looking at the proliferation or ingrowth of vascular cells from the vasculature into the sample and toxicity can be assessed by a decrease in cell number.
  • Figures 1A and IB are flow charts showing the steps for gel loading of an exemplary gel channel with one port to form a vascular matrix with one hole.
  • Figure 1A shows a side view of the gel channel during gel loading
  • Figure IB shows a top view of the gel channel during gel loading.
  • Figures 1C and ID are flow charts showing the steps for gel loading of an exemplary gel channel with three ports to form a vascular matrix with three holes.
  • Figure 1C shows a side view of the gel channel during gel loading
  • Figure ID shows a top view of the gel channel during gel loading.
  • Figures 2A-2F are diagrams showing structures of the vasculature bed device with a central well with or without inserted samples.
  • Figures 2A, 2C are diagrams showing a side view of the gel channel, Figure 2A is before and 2C is after spheroid or organoid deposition.
  • Figures 2B, 2D are diagrams showing a top view of the gel channel before (2B) and after (2D) spheroid or organoid deposition.
  • Figure 2E and 2F are the flow conditions that can be generated inside the tissue construct after samples are inserted into the central well.
  • Figure 2E is a diagram showing a side view of a device receiving an interstitial flow applied into the central port.
  • Figure 2F is a diagram showing the front view of a device that has luminal flows inside vasculatures.
  • Figures 3A-3F are diagrams showing steps in spheroid or organoid deposition in a vascular matrix with three holes and interstitial flow and lamina! flow through the spheroid or organoid.
  • Figures 3A, 3C, and 3E are diagrams showing a side view of the gel channel during spheroid or organoid deposition.
  • Figures 3B, 3D, and 3F are diagrams showing a top view of the gel channel during spheroid or organoid deposition.
  • Figure 4A is a diagram showing the direction of cell perfusion 90 and interstitial flow 92 through the vascular matrix with a spheroid or organoid 80.
  • Figure 4B is a diagram showing a quantification method based on vasculature and tumor spheroid.
  • Immune cells flowing inside the vascular networks extravasate and migrate toward the tumor spheroid 80. They can be regrouped into three categories: (1) immune cells that extravasated and migrated toward the tumor spheroid, the ones that are inside the center volume below the hole and infiltrate the tumor spheroid; (2) immune cells that extravasate but do not move toward the tumor spheroid; and (3) immune cells that stay luminal (within the vascular network).
  • Figure 5A is a cross-sectional presentation of a microfluidic device with three holes.
  • Figure 5B Schematic projection of a device that has interstitial flows applied from an open-top to generate fluid flow from a sample outward. Interstitial flows that pass by the sample can be generated by applying a pressure head on the sample’s compartment. The pressure head can be generated by a column of media or a pump.
  • Figure 5C Generation of interstitial flow from the organoid toward the vasculature using a pressure head by a hydrostatic media column.
  • FIG 6A The microfluidic chip (transparent) without cells.
  • the gel channel is sandwiched between two media channels and has an open-top well at the center.
  • Figure 6B Microfluidic device with tumor spheroid surrounded by vasculatures perfused with blue Dextran.
  • Figure 6C Fabrication method of the vascularized tumor tissue and monocyte recruitment assays.
  • perfusable vascular networks are created by seeding human umbilical vein endothelial cells (HUVECs) and normal human lung fibroblasts (NHLFs) in fibrin gels before inserting tumor spheroids made by triculture of MDA-MB-231 tumor cells, NHLFs and macrophages differentiated from bone-marrow derived monocytes, denoted 231 TFM.
  • HUVECs human umbilical vein endothelial cells
  • NHLFs normal human lung fibroblasts
  • FIG. 6D Experiment timeline of cell culture and imaging.
  • Figure 6E Top view of a region of a device that has one well containing a tumor spheroid and one empty well. Confocal images were recorded at day 0 and day 2 showing that in a device with tumor spheroid that contain 231 TFM recruits better monocytes from the vasculature (Ei, Eii) than device with MDA-MB-231 tumor cells and NHLFs co culture (231 TF) or control devices (Ctrl) that have only gel in the well.
  • Figure 6F Graphs showing the total number of monocytes migrating into the central well over the total number of monocytes in a 3x3mm region of interest of the well on day 2.
  • Figures 7A-7C Figure 7A is a timeline for the study of interstitial flow effect, macrophage polarization on T-cell recruitment using a device that has tumor spheroid embedded in fibrin well.
  • Figure 7B demonstrates that tumor spheroids do not recruit T-cells before interstitial flow is applied.
  • Figure 7C shows that T-cells are recruited into the spheroid hole in the presence of interstitial flow from the spheroid toward the vasculatures.
  • Figure 8A, 8B, 8C Figure 8Ai shows T-cell infiltration into the tumor spheroid compartment from the vasculature.
  • Figure 8AB is the zoom of the rectangle area in Figure 8Ai.
  • Figure 8Bi shows the cytotoxicity of tumor cells caused by infiltrating T-cells in the absence or presence of macrophages.
  • B16 OVA spheroid without macrophages (Figure 8Bi, Bii) display tumor cells killed by specific OT-1 T-cells, shown by Annexin V signal.
  • Figure 8Bii is the zoom of the rectangle area in Figure 8Bi.
  • Figure 9A is a prospective view of a device with two independent vasculature circuits for blood and lymphatic network co-cultures. An open-top channel is sandwiched between two other gel channels that have either blood or lymphatic vasculatures. All three gel channels are flanked by media channels.
  • Figure 9B is a prospective view of a mold for the fabrication of multiplexed devices.
  • Figure 9C is a cross- sectional view of section M-M of the device in Figure 9A after seeding vasculatures and depositing several layers containing skin cells such as epidermis containing keratinocytes and melanocytes, dermis containing dermal fibroblasts, and hypodermis containing adipocytes.
  • This 3D cell culture construct represents a skin model with several skin layers and perfusable vasculatures inside an open-top microfluidic device.
  • Figure 10A and 10B Figure 10A are z-stack images of different devices having vascularized tumor tissue for monocyte migration characterization. From left to right and top to bottom, overlap z-stack images on day 2 of: a control device without a tumor spheroid (only matrix inside the central well), a device having a 231 TF tumor spheroid, a device containing 2a 31 TFM tumor spheroid, a device containing a spheroid composed of MDA-MB-468, fibroblast and macrophages co-culture (468 TFM), a device containing a 231 TFM tumor spheroid treated with drug X or that treated with drug Y. These two drugs are two experimental antibodies that can block monocyte migration.
  • Figure 10B is a graph of chemotaxis coefficients of monocytes inside the hole compartment in different conditions in the devices shown in Figures 10A on day 2 and day 3.
  • Figure 11A displays two devices that have two tissue fragments of different weights, which are obtained from the same non-small lung cancer patient ex vivo tumoral tissues. Tumor tissues are fragmented into smaller pieces, before being reconstituted into a medium tissue sample (MTS) of 0.032+0.011 mg and a large tissue sample (LTS) of 0.2+0.048 mg. Perfused monocytes are recruited by the ex vivo tissue, proved by a strong presence of monocytes inside the central hole compartment at day 2.
  • Figure 11B show the chemotaxis coefficient of monocyte in the central hole calculated on day 2 and day 3. We can see that there are more monocyte migration inside the central hole in the device with MTS or LTS than that of a control device on both days.
  • Figure 12 Example of an angiogenesis assay. Image of a device with a 231 TF spheroid on day 0 and day 4. We observe that new microvasculature vessels sprout into the tumor spheroid by angiogenesis from the existing vasculatures.
  • Figure 13A Comparison of the permeability of lOkDa dextran in devices that have a presence or absence of an MDA-MB-468 TFM tumor spheroid.
  • Figure 13B Comparison of the permeability of 200nm-size nanoparticles in devices that have a presence or absence of an MDA-MB- 468 TFM tumor spheroid.
  • microfluidic refers to devices with dimensions of fluidic pathway elements for manipulating and controlling fluids, usually in the range of microliters (10 6 ) to picoliters (10 12 ).
  • the microfluidic devices typically include a channel or a portion of a channel with dimensions from tens to hundreds of micrometers
  • perfusable refers to a structure permitting the flow of fluid through vascular elements.
  • a perfusable tissue is a tissue having vascular elements crossing through the tissue and passing through the tissue.
  • Vascular elements include hollow structures, such as hollow lumens lined with endothelial cells, and including capillaries and other blood vessels, etc.
  • the vascular elements may include vascular networks.
  • the term “infiltrate” refers to cells or tissues passing through vascularized and non-vascularized tissue masses.
  • the infiltrate typically includes one or more of cells, extracellular matrix components, and exudate in an aqueous base.
  • Examples of infiltrate components are immune cells isolated from blood including T-cells, monocytes, natural killer cells, neutrophils, B-cells, and cells.
  • the infiltrate typically passes through the vascularized and non-vascularized tissue masses by movement around the cells of the vascularized and non-vascularized tissue masses, crossing any vascular elements, if present.
  • vascular network refers to a network of vascular elements, such as a network of hollow structures, hollow lumens lined with endothelial cells, capillaries blood vessels, etc.
  • vascular networks may be within tissue masses, as well as outside of tissue masses. Tissue masses containing vascular networks are typically perfusable tissue masses.
  • tissue masses refers to aggregates of cells self-assembling into three-dimensional masses. Tissue masses include tumors, spheroids, organoids, and other self-assembled masses. As used herein, “spheroids” typically refers to a cluster of cells from a cultured cell line. As used herein, “organoid” refers to cell clusters in extracellular matrix such as primary cells in an extracellular matrix (“ECM”). This can include tissues obtained by biopsy.
  • ECM extracellular matrix
  • Microfluidic platforms for forming and culturing perfusable vascularized tissue and/or perfusable vascularized tissue masses have been developed.
  • the first platform with fully-formed and perfusable vascular networks that have empty holes dedicated to host spheroids or organoids provides a platform for studies using an established vasculature for co culture with spheroids, organoids, cell monolayers, or cells in suspension. It also allows the perfusion of immune cells into the vasculature.
  • the open-top hole can be dedicated to generate flow that passes through the tumor spheroid toward the vasculature, mimicking the interstitial flow of tumor microenvironments .
  • the platforms typically include one or more microfluidic devices (also referred to as “microfluidic chips”).
  • the platform typically includes at least one microfluidic device.
  • the platform may include between about 2 and 3, 4, 5, 6, 8, 10 or 12 microfluidic devices.
  • the platform may include a single microfluidic device. Suitable sizes for the platform sides are between 5 mm and 280 mm.
  • the platform may have its sides with a length and width ranging between about 5 mm and 280 mm, between about 10 mm and 200 mm, between about 20 mm and 60 mm, between about 30 mm and 50 mm.
  • the platforms are the size of microscope slide cover glasses and are about 40 mm in length and 24 mm in width.
  • Each microfluidic device includes at least one gel channel.
  • the gel channel typically includes two ends and a top surface and a bottom surface.
  • the gel channel also includes one or more ports.
  • the one or more ports of the gel channel are positioned at the top surface of the device and at a distance away from the two ends of the gel channel.
  • the distance may be between about 1 mm and about 20 mm, such as between about 1 mm and about 15 mm, between about 1 mm and about 10 mm, between about 1 mm and about 5 mm, between about 5 mm and about 20 mm, such as between about 5 mm and about 15 mm, between about 5 mm and about 10 mm, or about 20 mm, or about 15 mm, or about 10 mm, or about 5 mm.
  • the microfluidic device typically includes openings connected to one or more columns or to tubing providing culture medium to the device. Fluid flow through the device channels is generally controlled by the hydraulic pressure of the culture medium and/or external pumps to generate the flows.
  • Endothelial cells and stromal cells are cultured in a hydrogel inside a hydrogel channel in a microfluidic device.
  • Representative hydrogels include MATRIGEL®, agar, hyaluronic acid, methyl cellulose, and other water swellable natural or synthetic polymers.
  • Within the hydrogel there is at least one empty space defined by the surrounding hydrogel and the bottom of the microfluidic channel. This empty space is connected to an open-top of the microfluidic device, forming a well. The gel channel is sandwiched between two media channels. Later, cells in the hydrogel form a network lined by endothelial cells that is also perfusable (Fig.
  • a cell-line spheroid or stem-cell-derived organoid or patient tissue-derived organoid or gels with cells in suspension is inserted.
  • These spheroids, organoids, or ex vivo tissues can also be termed as “inserted sample”.
  • An inserted sample is first suspended in a buffer solution and transferred to the well that is also filled with a buffer solution.
  • the inserted sample sinks to the bottom of the well.
  • the buffer solution in the well is then removed and the well is filled with a second hydrogel.
  • the hole is then connected to a fluidic pipeline to generate interstitial flow from the samples toward the vascular networks and side channels.
  • the hole can perform as reservoir for the inserted tissue’s specific media.
  • immune cells are perfused into the self-assembly perfusable microvascular tubes.
  • Immune cells can be primary immune cells isolated from blood, for examples: T-cells, monocytes, natural killer cells, neutrophils, B-cells, or cell lines. Immune cells are recruited by the samples that secrete various chemokines.
  • hydrogels that are used for the culture of microvasculature and sample can be originated from either the same or different matrix types. They are either natural extracellular matrix such as fibrin, collagen I, collagen IV, Fibronectin, laminin, vitronectin, D-lysine, MATRIGEL® (a mixture of collagen, basement membrane matrix components such as laminin, and extracellular matrix proteins), Heparan Sulfide proteoglycans or artificial polymers such as polyethylene glycol, or the combinations thereof.
  • natural extracellular matrix such as fibrin, collagen I, collagen IV, Fibronectin, laminin, vitronectin, D-lysine
  • MATRIGEL® a mixture of collagen, basement membrane matrix components such as laminin, and extracellular matrix proteins
  • Heparan Sulfide proteoglycans or artificial polymers such as polyethylene glycol, or the combinations thereof.
  • endothelial cells can be originated from primary endothelial cells from different organs such as umbilical vein endothelial cells, dermal vein endothelial cells, or differentiated from induced pluripotent stem cells, embryonic stem cells, or primary lymphatic endothelial cells from different organs.
  • Stromal cells are primary cells from the connective tissues such as fibroblasts, pericytes, astrocytes or cells that are differentiated from induced pluripotent stem cells or embryonic stem cells.
  • solutions are perfused through the microvasculatures are applied, either separately or simultaneously with interstitial flow from the sample ports.
  • the system can be used for drug screening.
  • chemical or biological drugs are introduced into the system.
  • Biological drugs can be therapeutic antibody or genetically-modified immune cells targeting any cell components of the 3D cell culture system, such as immune cells, vasculatures or cells inside the samples.
  • the sample is made by co-culturing tumor cells, connective tissue cells, immune cells or endothelial cells. They can form aggregates or solid tissues with a known composition.
  • the sample is an organoid obtained by differentiation of stem cells in a well plate or in a gel.
  • the sample is either animal tissue harvested from an animal or human tissue retrieved from a biobank or donated by a volunteering person.
  • an endothelial monolayer is present in the media channel and covers the media channel, as well as a side of the gel of the gel channel.
  • there are several cylindrical holes within the 3D cell culture device contain either similar or different types of samples or only gel without a sample which serves as a control.
  • the independent microfluidic circuits can be arranged in a multi array format to increase the throughput.
  • the multi-array device can be arranged in 6-, 12-, 14-, 48-, 96- 384- or 1536 devices in a rectangular matrix.
  • microvasculatures can mimic blood-brain barrier by culturing brain endothelial cells with astrocytes and pericytes.
  • the pre-vascularized device can be supplied to a research institute for integration of its samples.
  • This device has several hydrogel wells that are surrounded by perfusable vascular networks but without an inserted tissue and interstitial flow fluidic connector.
  • the pre-vascularized device is a 3D cell culture device including at least one microfluidic channel that has one or several empty wells formed by hydrogels that contain self-assembly 3D perfusable microvascular networks.
  • the cylindrical hole is dry and has no cells inside.
  • This pre-vascularized device can be used for the integration of its spheroids or cells into the empty wells.
  • an endothelial monolayer is present in the media channel and covers the media channel, as well as one or both sides of the gel in the gel channel.
  • a microfluidic device that has a gel channel having a cylindrical hole that contain samples, the gel channel is sandwiched between two other gel channels that contain lymphatic endothelial networks and blood endothelial networks respectively (Figure 9).
  • the device can be used to investigate the effect of various physical and biological components of the tumor microenvironment on immune cell recruitment.
  • this platform allows integration of tumor spheroids into the hole where they are immediately in contact with a vascular bed that is also perfusable.
  • the open-top hole is used to generate interstitial flows from the tumor spheroid toward the vasculature, mimicking the physiological interstitial flow emanating from a tumor.
  • tumor spheroids include mature M2 macrophages, they recruit more monocytes.
  • microfluidic device with one port in a gel channel is shown in Figures 1A and IB.
  • the microfluidic device typically includes a gel channel 20 with a port 10 and two media channels 26 and 28.
  • the two media channels 26 and 28 are positioned along the gel channel 20 and are separated from the gel channel 20 by phase guides 16 (See Figure 5A: Cross-sectional image of a 3-gel-hole device.
  • the microfluidic device channels 20, 26, and 28 typically include a bottom surface 22 and a top surface 24.
  • the bottom surface 22 may be formed of glass, while the top surface 24 may be formed of a polymer.
  • the port 10 is positioned on the top surface 24.
  • Typical dimensions for the channels which are preferably rectangular but may be round, are height between 200 and 500 microns, width between 5 mm and 1 cm, and length about 2 cm.
  • the gel channel has two open ends.
  • the port 10 of the gel channel 20 is typically positioned at a distance away from the channel’s two ends.
  • the distance may be between about 1 mm and about 20 mm, such as between about 1 mm and about 15 mm, between about 1 mm and about 10 mm, between about 1 mm and about 5 mm, between about 5 mm and about 20 mm, such as between about 5 mm and about 15 mm, between about 5 mm and about 10 mm, or about 20 mm, or about 15 mm, or about 10 mm, or about 5 mm.
  • a microfluidic device with one port in a gel channel is shown in Figures 1C and ID.
  • the microfluidic device with more than one port will include spacers in the ports.
  • a microfluidic device with a gel channel 30 may include three ports 10, 12, and 14, where port 12 includes a spacer 18a and port 14 includes a spacer 18b.
  • the spacers 18a and 18b typically match with their cross-sectional dimensions with the cross-sectional dimensions of the ports 12 and 14, for example, 0.5 to 5 mm.
  • the gel channel 30 is positioned between two media channels 36 and 38.
  • the two media channels 36 and 38 are positioned along the gel channel 30 and are separated from the gel channel 30 by phase guides 16.
  • the microfluidic device channels 30, 36, and 38 typically include a bottom surface 32 and a top surface 34.
  • the bottom surface 32 may be formed of glass, while the top surface 34 may be formed of a polymer.
  • the microfluidic devices and platforms are typically formed of inert polymers such as polydimethylsiloxane (PDMS) and polysulfone (PSF).
  • PDMS is a versatile elastomer that is easy to mold
  • PSF is a rigid, amber colored, machinable thermoplastic.
  • suitable materials include biologically stable thermosetting polymers, including polyethylene, polymethylmethacrylate, polyurethane, polyetherimide, polyimide, ultra-high molecular weight polyethylene (UHMWPE), cross-linked UHMWPE and members of the poly ary letherketone (PAEK) family, including polyetheretherketone (PEEK), carbon-reinforced PEEK, and polyetherketoneketone (PEKK).
  • thermosetting polymers include polyetherketoneketone (PEKK) and polyetheretherketone (PEEK). Glass can be used as a part of the device, bonded to the plastic layer to create the microfluidic channel. These can be made using conventional methods such as molding or extrusion, and bonding of the device to a substrate such as a glass coverslip.
  • PEKK polyetherketoneketone
  • PEEK polyetheretherketone
  • Methods of making microfluidic devices include stereolithography, soft lithography, laser machining, laser cutting, micromachining, micromilling, curing, bonding, 3D printing, molding, micromolding, and combinations thereof.
  • the top layer such as the top surface
  • the bottom layer such as the bottom surface
  • the top layer and the bottom layer are formed together through additive manufacturing.
  • the microfluidic devices may be formed to match the size of a cover glass to permit easy microscopic evaluation of tissues in the gel channel.
  • the platform may be formed of any size suitable to accommodate a desired number of microfluidic devices.
  • the platforms are useful to form in vitro perfusable vascular tissues and perfusable vascular tissue masses with infiltrates.
  • the gel is seeded with about 4-12 million endothelial cells/ml with or without stromal cells such as 0.5 to 2 million/ml fibroblasts, astrocytes, pericytes or adipocytes.
  • the perfusable vascular tissues are formed by the following process: a) Calculation of the precise gel solution volume that is equal to the gel channel volume minus the volume of the cylindrical gel holes. b) Injection of a gel solution containing cells into the microchannel from a vertical hole of the microfluidic chip. c) Pushing the gel further into the device using a bubble. Pins are removed from the wells when gels are still in a liquid state. d) The gel is kept from coming back to the hole due to surface tension of gel solution in the microfluidic channel. e) The hydrogel form of a hole is maintained until the gel solidification.
  • the methods include seeding a gel channel of the microfluidic platforms with a gel solution preferably containing cells via a port, gelling the gel solution to form a hole at the port, and initiating cell perfusion as well as interstitial flow through the vascular networks.
  • a spheroid or an organoid sample is inserted into the hole or port in the gel tube.
  • the formed in vitro perfusable vascular tissue can be used as an in vitro model for studying different diseases.
  • the formed in vitro perfusable vascular tissue masses with infiltrates may be used as in vitro models integrating tissue growth with immune system interactions, in vitro models for interaction between different tissue masses, as organ-on-a-chip or as human-on-a-chip.
  • Endothelial cells and stromal cells are cultured in a hydrogel inside the gel channel of the microfluidic device. Inside the hydrogel, there is at least one empty space defined by the surrounded hydrogel and the bottom of the microfluidic channel. This empty space is connected to an opening of the microfluidic device, forming a well. The gel channel is sandwiched between 2 media channels. Later, cells in the hydrogel form a network lined by endothelial cells that is also perfusable.
  • a cell-line spheroid or stem-cell-derived organoid or cells in suspension or patient tissue-derived organoid is inserted.
  • These spheroids, organoids or ex vivo tissues can also be termed as “inserted sample”.
  • An inserted sample is first suspended in a buffer solution then transferred to the well that is also filled with a buffer solution.
  • the inserted sample sinks to the bottom of the well.
  • the buffer solution in the well is then removed and the well is filled with a second hydrogel.
  • the hole is then connected to a fluidic pipeline to generate interstitial flow from the samples toward the vascular networks and side channels or inversely, from the vascular networks and side channels toward the samples.
  • Figures 1A is a flow chart showing the steps for gel loading of an exemplary channel with one port to form a vascular matrix with one hole.
  • Figure 1A shows a side view of the channel during gel loading.
  • Figure IB shows a top view of the channel during gel loading.
  • Gel loading method for the formation of a gel hole with well-defined dimensions of 0.5 to 5 mm typically includes the following steps.
  • gel solution 50 containing stromal and endothelial cells is loaded inside a pipette tip 52 and positioned on a port 10 of a microfluidic chip that has a central gel channel 20 and two media channels 26 and 28.
  • the gel solution 50 is pushed into the device.
  • the gel channel 20 is separated from the media channels 26 and 28 by a phase-guide 16 that keeps the gel solution inside the gel.
  • the gel solution 50 is confined inside the gel channel 20, which has a larger volume than the gel solution 50.
  • the volume of the gel solution 50 is precisely calculated by subtracting the gel volume under the port from the volume of a full gel channel 20.
  • the gel solution 50 is pushed further by generating a bubble at the port 10. As the gel solution 50 is pushed forward, the hydrophobic surface of the plastic becomes wet.
  • the surface tension generated by the gel channel keeps gel inside the channel and keeps the gel hole 40 empty and dry.
  • the hole 40 stays dried until gelification. Therefore, if there are some remaining cells within the hole 40, they will die out.
  • a perfusable vascular network lined by endothelial cells is formed due to paracrine secretion from fibroblasts.
  • the networks surround the hole 40 but do not grow into it.
  • the gel channel may include more than one port and the formed vascular networks may include more than one hole.
  • An exemplary method for loading these gel channels is shown in Figures 1C and ID.
  • Figures 1C and ID are flow charts showing the steps for gel loading of an exemplary channel with three ports to form a vascular matrix with three holes.
  • Figure 1C shows a side view of the channel during gel loading
  • Figure ID shows a top view of the channel during gel loading.
  • Gel loading method for the formation of multiple gel holes with well- defined dimensions typically includes the following steps. 1.
  • Gel solution 50 is loaded into the gel channel 30 that has several ports, such ports 10, 12, and 14. Beside the gel-loading port 10, two other ports 12 and 14 each have a spacer 18a or 18b inserted.
  • the gel solution is still liquid, therefore when the two spacers 18a and 18b are removed from the device, the gel solution 50 is first pulled back to the port caused by the local vacuum from the moving spacers, then moves back to the gel channel 30 due to surface tension.
  • Spacers may be a cylinder piece of metal or plastic that keeps the gel from flowing into the port during the loading process.
  • Figures 2A-2F are diagrams showing steps in spheroid or organoid deposition in a vascular matrix with one hole and interstitial flow and lamina! flow through the spheroid or organoid.
  • Figures 2A, 2C, and 2E are diagrams showing a side view of the channel during spheroid or organoid deposition.
  • Figures 2B, 2D, and 2F are diagrams showing a top view of the channel during spheroid or organoid deposition.
  • the method of spheroid or organoid deposition and interstitial flow may include the following steps. i. Spheroid or organoid deposition and interstitial flow in a device with one port
  • vascular networks 60 are formed in gelled gel solution 50 with one gel hole 40.
  • Spheroid or organoid 80 suspended in a matrix 82 of choice, are added to the hole 40. If the matrix 82 is too viscous, the spheroid/organoid 80 can be stuck during the loading process. Therefore, the spheroid/organoid 80 can be suspended in a buffer and added to the hole. The spheroid/organoid 80 will come down to the bottom of the hole by gravity where the networks are in proximity. The buffer will then be removed and replaced by the matrix 82 of choice.
  • Devices with multiple ports, and therefore, multiple gel holes may be used where one sample hole is used for accommodating a tumor spheroid/organoid, and other holes serve as controls that have only matrix or non-tumor cells or a monolayer. These devices may also be used in integrating multiple spheroids/organoids into one single vascular bed to study the interaction between multiple spheroids/organoids. This can be used to create a multi-spheroid/multi-organ platform or human-on- a-chip that also includes a vasculature.
  • Figures 3A-3F An exemplary device and spheroid or organoid deposition into the device are shown in Figures 3A-3F.
  • Figures 3A-3F are diagrams showing steps in spheroid or organoid deposition in a vascular matrix with three holes and interstitial flow through the spheroid or organoid and luminal flow.
  • Figures 3A, 3C, and 3E are diagrams showing a side view of the channel during spheroid or organoid deposition.
  • Figures 3B, 3D, and 3F are diagrams showing a top view of the channel during spheroid or organoid deposition.
  • vascular networks 60 are formed in gelled gel solution 50 with three gel holes 40, 42, and 44.
  • Spheroid or organoid 80 suspended in a matrix 82 of choice, are added to the hole 40. If the matrix 82 is too viscous, the spheroid/organoid 80 can be stuck during the loading process. Therefore, the spheroid/organoid 80 can be suspended in a buffer and added to the hole. The spheroid/organoid 80 will come down to the bottom of the hole by gravity where the networks are in proximity. The buffer will then be removed and replaced by the matrix 82 of choice.
  • the gel holes 42 and 44 may be used as controls and be filled with only matrix 84, or non-tumor cells, or a monolayer.
  • Two types of flow can be generated: (1) Interstitial flow from the top of the gel hole 40 (as well as from the top of the gel holes 42 and 44), toward the vascular networks. This flow is from the matrix-basal side of the endothelium toward the luminal side; and (2) luminal flows which are flows that can be generated by a pressure difference between two media channels across the vascular network 60.
  • Figure 4A is a diagram the direction of cell perfusion 90 and interstitial flow 92 through the vascular matrix with a spheroid or organoid 80.
  • Figure 4B is a diagram showing a quantification method based on vasculature and tumor spheroid.
  • Immune cells flowing inside the vascular networks extravasate and migrate toward the tumor spheroid 80. They can be regrouped into 3 categories: (1) immune cells that extravasated and migrated toward the tumor spheroid are the ones that are inside the center volume below the hole and infiltrate the tumor spheroid; (2) Immune cells that extravasate but did not move toward the tumor spheroid and; (3) immune cells that stay luminal.
  • Generation of interstitial flow and immune cell recruitment assay may use the following steps: flow may be applied from above the port of the device, passing across the extracellular matrix surrounding the spheroid and directed toward the vascular network.
  • the step of flowing medium through the gel channel typically includes the flow of culture medium through the channel controlled by hydraulic pressure applied to a culture medium.
  • the interstitial flow rates within the gel are between 0.001 to 10 pl/s.
  • the gel solution in the gel channel is incubated for a period between about 2 and about 10 days.
  • Perfusable vascular tissues and perfusable vascular tissue masses with infiltrates typically form after about 2 days, although it may take up to about 10 days in an in-vitro culture.
  • the spheroid or organoid may then be imaged through the hole or port in the gel channel in the microfluidic device to visualize the infiltrates ⁇ This is facilitated by the short distances through the port to the tissue.
  • Immune cells flowing inside the vascular networks extravasate and migrate toward the tumor spheroid. They can be regrouped into three categories: (1) immune cells that extravasated and migrated toward the tumor spheroid are the ones that are inside the center volume below the port and infiltrate the tumor spheroid; (2) immune cells that extravasate but do not move toward the tumor spheroid; and (3) immune cells that stay luminal (within the vascular network).
  • immune cells migrate into a proximity of the tumor spheroid, they can interact with the tumor cells. Cell interactions include direct contacts to the tumor cells, their killing by cytotoxicity, modifications in gene expression, and change in cell phenotypes and cellular compositions of the spheroid.
  • Figures 5A-5C are schematic projections of a device that has interstitial flows applied from an open-top to generate fluid flow outward from a sample. Interstitial flows that pass by the sample can be generated by applying a pressure head on the sample’s compartment. The pressure head can be generated by a column of media or a pump.
  • Figure 5A is a schematic presentation of the front of a microfluidic device with three gel holes as well as transverse and sagittal two cross-sectional views of the same device.
  • FIG. 5B is a diagram showing different compartment within the microfluidic device when interstitial flows are generated.
  • the pressure head can be generated by a column of media or a pump as shown in Figure 5C.
  • Figure 5C is a microfluidic device that has interstitial flows from the organoid toward the vasculature generated by a hydrostatic media column.
  • the platforms and devices can be used to establish in vitro models of diseases by replicating the inter-organ or inter-tissue interactions on one chip.
  • the established perfused tissue masses with infiltrates may be used for the screening of therapeutic agents targeted toward, or genetic factors relevant to, the diseases.
  • higher-magnification live imaging e.g., 30-60x
  • the technique is highly accessible to standard biology and engineering laboratories, conventional labware, a confocal microscope and standard cell culture equipment.
  • This device can also be used to establish a model of angiogenesis from a vascular network.
  • Live-imaging, alignment of 3D images and subtraction of thresholded images can be used to identify the difference between images, which quantify the volume of 3D angiogenic sprouting of vessels into the samples.
  • the devices are useful for studying the effects and/or effective dosage of therapeutic, prophylactic and/or diagnostic agents, for assessing an immunological role or effect on vascular structures, and for characterizing cellular interactions between the sample and the vascular network.
  • the agent(s)e can be applied directly to the sample or vascular network through the entry /sample port in the gel channel.
  • the effects of the agents can be assessed visually through the entry/sample port, by detecting and/or measuring cell viability, phenotype, cell migration or cell composition, cellular function (such as contraction of heart cells or production of insulin by islet cells) or changes in gene expression or products produced by the cells in response to the agent, or by other means of quantitating cells or cellular products or changes therein over time as a function of the agent.
  • angiogenesis can be assessed by looking at the proliferation or ingrowth of vascular cells into the sample and toxicity can be assessed by a decrease in cell number.
  • a 3D vascular cell culture model that allows the creation of an off-the-shelf in vitro human microvasculature containing hollow spaces for the insertion of spheroids or organoids. This is unique in terms of the device and method used to create the hollow space structures within a perfusable vascular network.
  • This system allows the study of immune-tumor cell interactions.
  • a tumor spheroid is surrounded by blood vessels, supporting extracellular matrix and stromal cells, similar to the in vivo human tumor microenvironment. With the support of perfusable networks surrounding the tumor, immune cells can flow into the blood vessels.
  • this technique Compared with the methodology used for other in vitro cell culture systems, this technique should better reproduce the complexity of a tumor by incorporating a perfusable microvasculature ⁇ Moreover, compared with the standard mouse xenograft model that uses human cells, this system offers high-resolution real-time imaging of tumor cell development and their interaction with immune cells within its complex microenvironment, while offering tight control of cell composition and physical parameters such as extracellular matrix composition and interstitial pressure.
  • Microfluidic devices contain gel inside a central compartment, which is flanked by channels containing cell culture media.
  • posts or a partial wall are used to keep gel confined within a channel while keeping in contact with cell culture media in adjacent channels.
  • an entry port for the gel solution loading is introduced and a new gel loading method, a method for generating a vascular bed with a defined morphology, interstitial and luminal flows and a new quantification method for cell migration are described. The method creates a well-defined 3D structure of gel inside a microfluidic device channel.
  • Prior art microfluidic devices have a central gel channel between two media channels.
  • the channel has a port from which a gel is injected that fills the channel underneath.
  • the gel is filled into the central gel region.
  • the gel is pushed further, and the gel is evacuated from the space underneath the port.
  • the gel is pushed further to generate a bubble that pushes the gel inside the hole away.
  • the pipette tip is removed, and the gel solution tends to regress back to the hole.
  • the gel solution does not move back up the port and is therefore held by the border of the port, leaving the gel adopting the form of the vertical hole ( Figure 1A).
  • Figure 1A illustrates the effect of capillary forces on maintaining the gel shape.
  • a capillary trap Using the principle of a capillary trap, one can make several gel holes within a gel slab by using spacers that block the gel from coming into a hole during gel loading. When the spacer is removed, it generates a vacuum that takes the gel coming back up the hole but is prevented from doing so by the capillary trap, thus allowing the gel to evacuate from the hole.
  • a successful device loading has an empty hole and media cannot travel into the hole. As the hole is dried, the vasculatures are perfusable and surround the hole but do not grow into the hole.
  • a 3D cell culture microdevice including at least one microfluidic channel containing a vascularized network in a hydrogel, having a hole or port, is used to form a cell-line spheroid, stem-cell-derived organoid, cells in suspension or patient tissue-derived organoid (also termed “samples”).
  • the 3D perfusable microvascular networks are created by co culturing endothelial cells with stromal cells inside a hydrogel that forms at least one cylindrical hole, the cylindrical hole is filled with a second hydrogel that contains sample(s).
  • the sample(s) can receive media from the top port through a fluidic pipeline to generate interstitial flow from the samples toward the vascular networks and side channels.
  • the microvascular network is formed from cells such as endothelial cells are originated from umbilical vein endothelial cells, induced pluripotent stem cells, lymphatic endothelial cells.
  • the microfluidic chip may be continuous or discontinuous perfusion flows through the vasculatures are applied, either separately or simultaneously with interstitial flow from the sample ports.
  • the 3D cell culture device includes an endothelial monolayer in the media channel(s), which covers the media channel, as well as the side of the gel channel.
  • there are several cylindrical holes the cylindrical holes containing similar or different types of samples or control gel without a sample.
  • the multi-array device can be arranged in 6-, 12-, 14-, 48-, 96- 384- and 1536 devices in a rectangular matrix.
  • the device can be used for screening chemical or biological agents applied to treat the cell sample.
  • the agents can be therapeutic, prophylactic and/or diagnostic.
  • One advantage of this system is that the vasculatures can mimic blood-brain barrier by culturing brain endothelial cells with astrocytes and pericytes.
  • biological drugs include therapeutic antibodies targeting immune cells, vasculatures or samples.
  • the 3D cell culture device can be used for testing of specific cell types, such as where the sample is made by co-culturing tumor cells, connective tissue cells, immune cells or endothelial cells. These may be in the form of aggregates or solid tissues with a known composition.
  • the sample can be an organoid obtained by differentiation of stem-cells in a well plate or in a gel.
  • the sample can be cells in suspension in media or hydrogels.
  • the sample can be ex vivo tissue that is either animal tissue harvested from an animal or human tissue retrieved from a biobank, obtained by biopsy, or donated by a volunteering person.
  • the sample can be deposited either by a pipette or automatically by a bioprinter nozzle.
  • the system can be used for a variety of screens or testing, for example, to characterize immune cell recruitment by the samples in the devices, by the quantification of immune cells perfused into the vascular networks that leave the blood vessels and migrate into the extracellular matrix containing the sample and interact with the sample, where the interaction include direct contact to the samples cells, their killing by cytotoxicity, modifications in gene expression, and/or change in cell phenotype and cellular composition.
  • the device is used in a process for quantification of 3D angiogenesis by time-lapse imaging of the device. At several time points, images are aligned and thresholded images subtracted to get the difference between images, which quantifies the volume of 3D angiogenic sprouting of vessels into the samples.
  • Example 1 Formation of Tumor Microenvironment for Testing
  • the gel solution had gel precursors and endothelial cells (for example, dermal, umbilical, and/or brain endothelial cells) and stromal cells (for example, fibroblasts, pericytes, astrocytes) in suspension in a fibrin gel solution and was injected into the device as described in Figures 1A and IB. Later, endothelial cells created a blood- vessel-like network that was also perfusable. The hole remains dried, without media, and surrounded by a vascular network. The blood vessel network structure surrounded the empty space in the hole and adopted a form similar to the hole.
  • endothelial cells for example, dermal, umbilical, and/or brain endothelial cells
  • stromal cells for example, fibroblasts, pericytes, astrocytes
  • a tumor spheroid was placed inside this hole, as shown in Figures 2A-2F.
  • the tumor is suspended in collagen or a mixture of collagen and fibrin, or MATRIGEL®.
  • Ex vivo tissues from a patient or an animal can also be incorporated into the gel hole within the vascular bed.
  • This system adds further complexity whereby the fluidic connection is sealed and a fluid flow is applied from the top of the spheroid to the media channel.
  • Generating fluid flow from the top of the spheroid toward the media channels recapitulates interstitial flows, which is a benchmark of several solid tumors.
  • Multi-hole designs ( Figures 3A-3F) have the advantage of being able to integrate controls to the experiment.
  • organoids can be integrated into a vasculature bed to study the interaction between organoids.
  • This platform can be further developed toward a vascularized human-on-a-chip model in which several organoids (including brain, heart, kidney, etc.) are connected by a vascular network.
  • Endothelial cells were cultured within the gel, which created 3D blood vessel structures lined by endothelial cells. This 3D blood vessel structure allowed the perfusion of immune cells.
  • the flow coming from the inlet on top of the tumor spheroid supported immune cells as they extravasated and migrated toward the spheroid by chemotaxis. This event was characterized quantitatively.
  • the immune cell populations were divided based on their relative position to the blood vessels and the central hole. By quantifying the number of immune cells that were inside the central hole, the percentage of the cells that extravasated and migrated in the gel among all cells in the region of interest was characterized.
  • the platform can be used to study the effects of tumor-associated macrophage within a tumor spheroid on monocyte recruitment.
  • a polydimethylsiloxane device with several vertical holes on top of the gel channel was used to create a perfusable vascular network.
  • the microfluidic chip is illustrated in Figure 6A and the tumor tissue with perfusable vasculature is illustrated in Figure 6B.
  • a fibrin gel solution that has HUVECs and NHLFs in suspension was injected into the device. These form a blood- vessel-like network that is perfusable (Figure 6C).
  • Tumor spheroids were formed by coculturing MDA-MB- 231 tumor cells (T), NHLFs (F), with or without non-polarized macrophages (M0) differentiated from bone marrow-derived monocytes in a low-adhesion 96 well-plate.
  • TFM MDA-MB- 231 tumor cells
  • M0 non-polarized macrophages
  • 231 TFM The spheroid obtained from the tri-culture of MDA-MB- 231 tumor cells, fibroblasts and macrophages
  • 231TF the one obtained from the co-culture of MDA-MB- 231 tumor cells and fibroblasts.
  • Monocytes extravasate and migrate more readily from the vasculatures into the gel well that contains a triculture 231 TFM tumor spheroid at day 2 (Figure 6Eii) than day 0 ( Figure 6Ei) and than the tumor spheroid does not have macrophages (231 TF, Figure 6Eiii) at day 2 or control devices without a tumor spheroid (Figure 6Eiv) at day 2.
  • the ratio of the number of monocytes inside the central hole to the total number of monocytes inside the region of interest of 3x3mm confirms higher migration into the hole on day 2 ( Figure 6F).
  • tumor spheroids were made by co culturing mouse B16-OVA melanoma cells, normal human lung fibroblasts and inserting them into a perfusable HUVEC network. OT-1 T-cells were then perfused into the network.
  • Tumor spheroids were made by co culturing mouse B 16-OVA melanoma cells, normal human lung fibroblasts and inserting them into a perfusable HUVEC network. OT-1 T-cells were then perfused into the network.
  • Figure 7 A shows the timeline for the study of interstitial flow and macrophage polarization effects on T-cell recruitment using the device having tumor spheroid embedded in fibrin well. Tumor spheroids do not recruit T-cells before an interstitial flow is applied (Figure 7B). T-cells are recruited into the spheroid hole in the presence of interstitial flow from the spheroid toward the vasculatures ( Figure 7C). T cell infiltration into the tumor spheroid compartment at day 2 is also observed in Figure 8Ai and Aii.
  • Extravasated OT-1 T-cells specifically killed B 16-OVA melanoma cells, as characterized by Annexin V apoptosis staining ( Figure 8Bi and Bii).
  • Figure 8Biii When Raw 264.7 macrophages are present in the tumor spheroid co-culture ( Figure 8Biii), fewer OT-1 T-cells infiltrate the tumor spheroid, and fewer tumor cell killing events are detected by Annexin V apoptosis staining ( Figure 8C).
  • Example 4 Co-cultures of blood endothelial networks and lymphatic endothelial networks to create a model of vascularized skin
  • Figure 9A is a design of the device with two independent vasculature circuits for blood and lymphatic network co-cultures.
  • the central channel has an open top and is sandwiched between two other gel channels that have either blood or lymphatic vasculatures.
  • the open-top compartment sandwiched by the endothelial and lymphatic compartment receives gel deposition of several layers of skin such as hypodermal layer consisted mainly of adipocytes, the dermal layer consisted mainly of normal human lung fibroblasts, and the epidermal layer consisted of a mix of melanocytes and keratinocytes. All skin layers are suspended in collagen or gelatin-based hydrogel.
  • Figure 9B is the design of a mold for the fabrication of multiplexed devices.
  • Figure 9C shows an example of the cross-section M-M when vasculatures are formed and then several layers of skin are deposited into the open-top channel of the device.
  • Example 5 Vascularized tumor model as a platform for screening therapeutic antibodies
  • Example 2 The system of Example 2 was used to form a vascularized tumor model to study the effect of different macrophage phenotypes on monocyte recruitment and screen antibody drugs that block monocyte recruitment by a tumor spheroid. Different vascularized tumor tissue devices are imaged on day 2.
  • the results show discrepancies of monocyte recruitment response (Figure 10A).
  • Devices having 231 TFM tri-culture display the highest monocyte recruitment, as more monocytes are present in the spheroid well, than under other conditions: control device without a tumor spheroid, device having a 231 TF tumor spheroid, spheroid composed of MDA-MB-468, fibroblast and macrophages co-culture (468 TFM), 231 TFM tumor spheroid treated with experimental antibodies that can block monocyte migration.
  • Chemotaxis coefficients of monocytes inside the hole compartment in different conditions in A on day 2 and day 3 were calculated, as shown in Figure 10B.
  • Chemotaxis coefficient represents the migration speed of monocytes under a chemoattractant gradient caused by the presence of tumor spheroid. Most monocyte migration happens on day 2 instead of day 3.
  • the tumor cell phenotype affects the capability of the tumor spheroid to recruit monocyte.
  • Antibody-drug efficacy can be evaluated by the assessment monocyte chemotaxis coefficient inside the tumor spheroid compartment.
  • Example 6 Monocyte recruitment by ex vivo patient tissues
  • Tumor tissues dissected from a non-small lung cancer patient were fragmented into smaller pieces, before being reconstituted into a medium tissue sample (MTS) of 0.032+0.011 mg and a large tissue sample (LTS) of 0.2+0.048 mg.
  • MTS medium tissue sample
  • LTS large tissue sample
  • a 231 TF spheroid was added into the device using two different media for the tumor spheroid and vasculature.
  • the 231 spheroid was fed with RPMI with 10% FBS and the vasculature was flanked in Vasculife with 2% FBS. This gradient of FBS helped new vessels form and sprout into the tumor spheroids.
  • Example 8 Measure molecular and nanoparticle permeability of the vascular networks
  • Fluorescent molecules and nanoparticles were perfused into the vascular networks. Time-lapse imaging was used to record the diffusion of these molecules and nanoparticles. This was used to compute the permeability of the vascular networks in the presence of an MDA-MB-468 tumor-fibroblast-macrophage tri-culture (468TFM) spheroid.
  • 468TFM tumor-fibroblast-macrophage tri-culture
  • Example 9 Freezing and thawing devices for transportation:
  • vascular network device To freeze devices for long-distance transportation, media inside a vascular network device was replaced with a commercial freezing media and the device placed inside a thermal insulation box at 4°C for 1 to 4 hours before transferring the box to a -80°C freezer overnight.
  • the device can be transferred to a liquid nitrogen tank or placed into dried ice.
  • the device On the day of use, the device is thawed at 37 °C and media perfused to revive cells inside the device.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Genetics & Genomics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Sustainable Development (AREA)
  • Cell Biology (AREA)
  • Dentistry (AREA)
  • Environmental Sciences (AREA)
  • Dispersion Chemistry (AREA)
  • Clinical Laboratory Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Vascular Medicine (AREA)
  • Apparatus Associated With Microorganisms And Enzymes (AREA)

Abstract

Dispositifs microfluidiques avec des orifices ouverts et des canaux de gel pour former des réseaux vasculaires d'hydrogel perfusables avec des trous ou des orifices pour des échantillons, procédés de fabrication et d'utilisation, intégrant des flux interstitiels à un modèle de tissu vascularisé ex vivo. Des échantillons cellulaires, de sphéroïdes, d'organoïdes et de tissus peuvent être utilisés pour le dépistage d'agents en matière d'efficacité, de toxicité et de dosage. Les dispositifs créent un flux interstitiel depuis le haut du trou de gel, à travers l'échantillon vers les réseaux vasculaires, et/ou des flux luminaux générés par une différence de pression entre deux canaux de milieu à travers le réseau vasculaire. Ce système est utile pour étudier l'angiogenèse, la migration des cellules immunitaires et pour tester de nouveaux candidats médicaments en immunothérapie.
PCT/US2022/017505 2021-04-23 2022-02-23 Plates-formes fluidiques pour tissus vascularisés perfusables avec infiltrations WO2022225602A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163179006P 2021-04-23 2021-04-23
US63/179,006 2021-04-23

Publications (1)

Publication Number Publication Date
WO2022225602A1 true WO2022225602A1 (fr) 2022-10-27

Family

ID=80735656

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/017505 WO2022225602A1 (fr) 2021-04-23 2022-02-23 Plates-formes fluidiques pour tissus vascularisés perfusables avec infiltrations

Country Status (2)

Country Link
US (1) US20230146860A1 (fr)
WO (1) WO2022225602A1 (fr)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116478819B (zh) * 2023-06-20 2023-09-26 清华大学 一种用于构建三维器官微环境模型的微流控系统及其制备方法和应用

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017070542A1 (fr) * 2015-10-22 2017-04-27 The Trustees Of The University Of Pennsylvania Systèmes et procédés de production de modèles micro-façonnés du col de l'utérus humain
WO2017155399A1 (fr) * 2016-03-09 2017-09-14 Mimetas B.V. Structures tubulaires doubles
US20180327701A1 (en) * 2015-11-06 2018-11-15 Universidad De Zaragoza Device and microfluidic system for studying cell cultures
WO2019200034A1 (fr) * 2018-04-11 2019-10-17 Georgia Tech Research Corporation Moelle osseuse-sur-puce, puce in vitro, multi-niche
US20200063081A1 (en) 2016-06-15 2020-02-27 Mimetas B.V. Cell culture device and methods

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017070542A1 (fr) * 2015-10-22 2017-04-27 The Trustees Of The University Of Pennsylvania Systèmes et procédés de production de modèles micro-façonnés du col de l'utérus humain
US20180327701A1 (en) * 2015-11-06 2018-11-15 Universidad De Zaragoza Device and microfluidic system for studying cell cultures
WO2017155399A1 (fr) * 2016-03-09 2017-09-14 Mimetas B.V. Structures tubulaires doubles
US20200063081A1 (en) 2016-06-15 2020-02-27 Mimetas B.V. Cell culture device and methods
WO2019200034A1 (fr) * 2018-04-11 2019-10-17 Georgia Tech Research Corporation Moelle osseuse-sur-puce, puce in vitro, multi-niche

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
AYUSO ET AL., ONCOIMMUNOLOGY, vol. 8, no. 3, 2019
BENNINGER ET AL., CURR PROTOC CELL BIOL., vol. 4, no. 11, 2013, pages 24
BHATIA ET AL., NAT. BIOTECHNOL., vol. 32, 2014, pages 760 - 772
BOUSSOMMIER-CALLEJA ET AL., BIOMATERIALS, vol. 198, 2019, pages 180 - 93
BOUSSOMMIER-CALLEJA ET AL., TRENDS IN CANCER, vol. 2, no. 1, 2016, pages 6 - 19
CAMPISI ET AL., BIOMATERIALS, vol. 180, 2018, pages 117 - 29
CHEN ET AL., INTEGR BIOL QUANT BIOSCI FROM NANO TO MACRO, vol. 5, no. 10, 2013, pages 1262 - 71
CHEN ET AL., NAT PROTOC., vol. 12, no. 5, 2017, pages 865 - 80
JAIN, NAT. MED., vol. 9, 2003, pages 685 - 693
NASHIMOTO ET AL., BIOMATERIALS, vol. 229, 2020, pages 119547
PAEK ET AL., ACS NANO, vol. 13, no. 7, 2019, pages 7627 - 43
PHILIPP ET AL., SCI REP, vol. 9, 2019, pages 9532
SHELTON ET AL., ISCIENCE, vol. 24, no. 1, 2021, pages 101985
SONTHEIMER-PHELPS ET AL., NAT REV CANCER, vol. 19, 2019, pages 65 - 81
WANG ET AL., MICROMACHINES, vol. 9, no. 493, 2018, pages 1 - 26

Also Published As

Publication number Publication date
US20230146860A1 (en) 2023-05-11

Similar Documents

Publication Publication Date Title
Fetah et al. The emergence of 3D bioprinting in organ-on-chip systems
Moya et al. In vitro perfused human capillary networks
Nashimoto et al. Integrating perfusable vascular networks with a three-dimensional tissue in a microfluidic device
CN109804057B (zh) 细胞培养装置以及细胞培养方法
Vickerman et al. Design, fabrication and implementation of a novel multi-parameter control microfluidic platform for three-dimensional cell culture and real-time imaging
EP2526978B1 (fr) Dispositif et procédé de génération de vaisseaux sanguins in vitro
Banerjee et al. Strategies for 3D bioprinting of spheroids: A comprehensive review
CA2720261C (fr) Plate-formes microfluidiques tridimensionnelles et procedes d'utilisation de celles-ci
Bonvin et al. A multichamber fluidic device for 3D cultures under interstitial flow with live imaging: development, characterization, and applications
Sato et al. Recent progress in the development of microfluidic vascular models
US20240139378A1 (en) Bioengineered vascular network
Sakthivel et al. Microfluidic analysis of heterotypic cellular interactions: a review of techniques and applications
US20230146860A1 (en) Fluidic platforms for perfusable vascularized tissues with infiltrates
Tan et al. Engineering strategies to capture the biological and biophysical tumor microenvironment in vitro
KR102395801B1 (ko) 뇌혈관 모사용 미세유체 디바이스 및 이를 포함하는 고효율 혈액뇌관문 모사 시스템
JP6296620B2 (ja) 細胞評価用ハイドロゲル基材、細胞評価用ハイドロゲル基材の作製方法および細胞評価手法
Kim et al. Microfluidic vascular models of tumor cell extravasation
Seaman et al. Recent advances in cancer-on-a-chip tissue models to dissect the tumour microenvironment
Barbato et al. A permeable on-chip microvasculature for assessing the transport of macromolecules and polymeric nanoconstructs
Hwangbo et al. Tumor-on-a-chip models combined with mini-tissues or organoids for engineering tumor tissues
Ravi et al. Engineered 3D ex vivo models to recapitulate the complex stromal and immune interactions within the tumor microenvironment
US20220284832A1 (en) Models and methods to establish perfused vascularized tissues in three-dimensional in vitro culture
US20230174909A1 (en) Cell culture system for perfusable networks of self-assembled cells
KR101898093B1 (ko) 생체관 모사조직이 포함된 분석용 칩 및 이의 제조방법
Huang et al. Microfluidic channel with embedded monolayer nanofibers for cell culture and co-culture

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22709891

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22709891

Country of ref document: EP

Kind code of ref document: A1